WO2004042041A1 - Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie - Google Patents

Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie Download PDF

Info

Publication number
WO2004042041A1
WO2004042041A1 PCT/AU2003/001476 AU0301476W WO2004042041A1 WO 2004042041 A1 WO2004042041 A1 WO 2004042041A1 AU 0301476 W AU0301476 W AU 0301476W WO 2004042041 A1 WO2004042041 A1 WO 2004042041A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
virus
nucleic acid
cell
subject
Prior art date
Application number
PCT/AU2003/001476
Other languages
English (en)
Inventor
Geoffrey Philip Symonds
Susan Margaret Pond
Gregory Charles Fanning
Original Assignee
Johnson & Johnson Research Pty Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Johnson & Johnson Research Pty Limited filed Critical Johnson & Johnson Research Pty Limited
Priority to JP2004548938A priority Critical patent/JP2006505259A/ja
Priority to AU2003275767A priority patent/AU2003275767A1/en
Priority to US10/534,135 priority patent/US20060051866A1/en
Priority to CA002505379A priority patent/CA2505379A1/fr
Priority to EP03810335A priority patent/EP1558723A4/fr
Publication of WO2004042041A1 publication Critical patent/WO2004042041A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/464838Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to cell and gene therapy, particularly as applied to hematopoietic cells of the Cytotoxic T Lymphocyte (CTL) class, either alone or in combination with exogenous gene-containing hematopoietic cells of the CD4+ T- lymphocyte class and /or the hematopoietic progenitor (HP) cell type.
  • CTL Cytotoxic T Lymphocyte
  • HP hematopoietic progenitor
  • adoptive immunotherapy aims to use the patient's cells to increase production of cytotoxic T lymphocytes (CTLs) to treat cancer or infection.
  • CTLs cytotoxic T lymphocytes
  • This technique has shown promise as a potential clinical treatment regime for human patients (Heslop et al, 1996; Walter et al, 1995).
  • the process works most effectively when the proper epitopes for presentation to the CTLs (this can be likened to an ex vivo "education" process of the CTLs) are known or can be identified.
  • Adoptive immunotherapy preferably also requires the presence of antigen presenting cells (APCs) that are capable of expressing at least one disease-specific epitope.
  • APCs antigen presenting cells
  • Current methods are now known for the efficient production of APCs and include, but are not limited to, International Publication PCT/US02/05748.
  • APC systems have been used to generate antigen-specific CTLs to a single epitope, including: 1) human dendritic cells (DC) pulsed with defined peptides; 2) peripheral blood mononuclear cells (PBMCs) which have been driven to lymphoblasts and pulsed with peptides; 3) lymphoblastoid cell lines (LCL) where the natural peptides are acid-stripped and loaded with the peptides of interest; 4) Drosophila cells engineered to express empty class I molecules; and Mouse 3T3 cells transfected with human class I and co-stimulatory molecules (Latouche and Sadelain, 2000).
  • DC dendritic cells
  • PBMCs peripheral blood mononuclear cells
  • LCL lymphoblastoid cell lines
  • CTLs can be produced to exhibit peptide-specificity to several HLA-A2.1-restricted peptides from melanoma-associated antigen.
  • These CTLs have been isolated from leukopheresis samples and presented in vitro with melanoma antigenic peptide epitopes using Drosophila cells as the non-natural antigen-presenting cells (nnAPCs).
  • nnAPCs non-natural antigen-presenting cells
  • the CTLs are expanded over a period of 20 or 21 days and incubated with autologous monocytes APCs loaded with the melanoma antigenic epitopes in the presence of Interleukin-2 (IL- 2) and Interleukin-7 (IL-7). This treatment is followed by non-specific expansion using OKT3 for 10 days and the product is infused into patients.
  • IL-2 Interleukin-2
  • IL-7 Interleukin-7
  • hematopoietic progenitor cells are particularly sensitive to HTV infection.
  • Therapeutic genes have been introduced into CD34+ pluripotent hematopoietic progenitor cells to target HIV gene expression.
  • Hematopoietic progenitor cells may be readily separated from more mature hematopoietic cells by using the CD34+ antigen.
  • the antigen is a membrane-bound 115 Kd molecule present on cells but absent on mature hematopoietic cells.
  • CD 34+ cells can give rise to multi-lineage colony forming cells, (Baum et al.
  • CD4+ and CD8+ T- lymphocytes are capable of relatively rapidly (3-6 months) reconstituting lymphoid (CD4+ and CD8+ T- lymphocytes) and myeloid (monocyte/macrophages) hematopoiesis through a series of intermediate cells of increasing maturity (Levinsky 1989; Schwartzberg et al. 1992).
  • CD8+ lymphocytes may be candidates for gene therapy ex vivo; however the ability of transduced CD8+ cells to target HIN nucleic acid in the body is not known.
  • CD4+ and CD8+ T-lymphocytes may be separated from other cell types by means of the CD4 and CD8 receptor respectively.
  • Cytotoxic T-lymphocytes CTLs are a sub-set of CD8+ T-lymphocytes that are involved in cellular response to infections and malignancies (Janeway et al, 1999).
  • CD4+ T lymphocytes are involved in T-lymphocyte helper function for B cells (antibody production - Janeway et al, 1999) and helper function for CD8+ T-lymphocytes.
  • Ribozymes are small catalytic RNA moieties capable of cleaving specific RNA target molecules. Ribozymes have been used to target a number of nucleic acid sequences.
  • ribozymes directed against HTV-1 can interfere with HIV-1 replication by interfering in several steps in the HTV-1 life cycle including the production of genomic viral RNA in recently infected cells (prior to reverse transcription) and the production of viral RNA transcribed from the provirus before translation or prior to genomic RNA packaging (Sarver et al. 1990; Sun et al. 1996; Sun et al. 1998).
  • ribozymes are more effective than antisense in their ability to inhibit gene expression because ribozymes are catalytic molecules that not only bind to their target but cleave their target.
  • ribozymes can cleave multiple RNA substrate molecules (Sarver et al. 1990; Sun et al. 1996).
  • RNA Ribonucleic acid
  • GUX target motif or, in certain cases, NUX may suffice (where N is any ribonucleotide and X is A, C or TJ ribonucleotides).
  • catalytic RNAs are unlikely to provoke an immune response that leads to the elimination of cells that contain the exogenous gene.
  • ribozyme cleavage activity in test tube reactions, and protective effects in tissue culture systems against laboratory and clinical isolates of HTV-1 (Sarver et al. 1990; Sun et al. 1998; Wang et al. 1998). These studies used either hammerhead or hairpin ribozymes.
  • a hammerhead ribozyme directed against a highly conserved region of the tat gene is provided in Figure 1 as Rz2.
  • the tat gene is essential for HTV-1 replication in that it encodes and produces the Tat protein.
  • the Tat protein is a transcriptional activator of integrated HTV-1 provirus.
  • the Rz2 complementary hybridizing and target sequences comprise nucleotides 5833-5849 (GGAGCCA GUA GAUCCUA) of reference strain HTV-HXB2 (Genbank accession number K03455) or nucleotides 5865 to 5882 (GGAGCCA GUA GAUCCUA) of HTV ILTB (Genbank accession number X01762).
  • the Rz2 ribozyme sequence 5 -TTA GGA TCC TGA TGA GTC CGT GAG GAC GAA ACT GGC TC-3' has been inserted as DNA into the 3' untranslated region of the neJ * gene within the plasmid pLNL6, which contains the replication-incompetent retroviral vector LNL6 (Genbank accession number M63653) to generate a new virus, RRz2.
  • the ribozyme sequence has been expressed as a i?eo R -ribozyme fusion transcript from the Moloney Murine Leukemia Virus (MoMLV) Long Terminal Repeat (LTR) in RRz2 as disclosed previously (Sun, L.-Q. et al. (1995) Proc.
  • the ribozyme gene transfer product RRz2 has been used in two Phase I Clinical
  • the RRz2 construct was introduced into a population of CD4+ T-lymphocytes (from the HTV negative twin) ex vivo and these cells (in a background of non-gene containing T-lymphocytes) were infused into the corresponding HTV positive twin. Subsequent to infusion, ribozyme construct presence and expression in mature lymphoid cells was seen for at least 4 years (the latest time point examined). This Phase I study has shown that this approach is technically feasible and safe.
  • the RRz2 construct was introduced into CD34+ progenitor cells ex vivo.
  • the infusion of RRz2-containing CD34+-HP cells gave rise to RRz2-contairting peripheral blood cells including CD4+ and CD8+ T-lymphocytes, as determined by sensitive PCR methods.
  • Ribozyme construct presence and expression was seen in both mature lymphoid and myeloid cells for up to 3.5 years (the latest time point examined).
  • Anti-retroviral drugs are used in the treatment of HTV/ AIDS. These drugs predominantly target the reverse transcriptase and protease steps of the HTV life-cycle. As of November 2002 there are of the order of 15 drugs that are in clinical use for the treatment of HTV/ AIDS (Guidelines for the Use of Antiretroviral Agents in HTV-Tnfected Adults and Adolescents, 2002; http:/ / www.hivatis.org). Generally two nucleoside and one non-nucleoside reverse transcriptase inhibitors or two nucleoside reverse transcriptase and one protease inhibitor are the initial drug combinations of choice.
  • treatment interruptions see guidelines for the Use of Antiretroviral Agents in HIV-Infected Adults and Adolescents, 2002; http:/ / www.hivatis.org.
  • treatment interruptions may be relatively short (several weeks) or of more prolonged duration (several months) and the safety of the Treatment Interruptions is monitored by assessment of viral load and CD4+ T-lymphocyte count, along with other safety measures.
  • a method for producing a cytotoxic T-lymphocyte population primed for virus-specific CTL activity comprising the steps of: (a) preparing non-naturally occurring antigen-presenting cells(nnAPC) which present at least one virus-specific antigen;
  • step (c) incubating a population of CD8+ cells obtained from the white blood cells in step (b) with the nnAPC cells; and (d) treating the CD8+ cells with one or more supportive cytokines.
  • a method for producing a cytotoxic T-lymphocyte population transduced with virus-inhibiting nucleic acid and primed for virus-specific CTL activity comprising the steps of: (a) preparing a non-naturally occurring antigen presenting cell line
  • nnAPC which presents at least one virus specific antigen
  • step (f) incubating the CD8 + cells with non-proliferating peripheral blood mononuclear cell-derived adherent cells and wherein the adherent cells present at least one of the same virus-specific antigens of step (a).
  • the present invention relates in one embodiment, to the use of cytotoxic T- lymphocytes (CTLs) that have been specifically exposed, and are sensitized, to viral disease specific peptide epitopes ex vivo for the purpose of eradicating diseased cells when those CTLs are re-infused into a patient. Further, the invention relates in another embodiment, to genetically modifying at least a percentage of the sensitized CTLs so that they are protected or are resistant to viral disease. In yet another form, the first mentioned embodiment may be used alone or in combination with the second mentioned embodiment above. In still another preferred embodiment, the virus-specific CTLs, which may comprise a percentage of gene modified CTLs, can be used alone or in combination with other hematopoietic cells, a proportion of which are gene-modified with disease-specific gene(s).
  • CTLs cytotoxic T- lymphocytes
  • the viral disease-specific cytotoxic T lymphocytes can be used for the treatment of infectious diseases, and further transduction of the cells with a disease protective genetic agent may further help in this treatment.
  • CTLs cytotoxic T lymphocytes
  • These transduced CTLs can be used alone or in combination with other cell populations, namely CD4+ and CD34+ cell populations that have been transduced with a therapeutic (or marker) gene construct for effect on disease. This approach will be useful for therapy of HTV-1 and for other cell/gene therapies in which protected blood cell populations are required.
  • the invention further relates to the use of hematopoietic cells, preferably transduced ex vivo (for example, with an anti-HTV-1 gene therapeutic), to be introduced into a recipient patient alone or in combination with other such hematopoietic cells in sufficient number to produce a chimeric hematopoietic system comprising enough disease specific (and in one embodiment anti-HIV agent-containing hematopoietic cells) targeting moieties to have a therapeutic effect.
  • hematopoietic cells preferably transduced ex vivo (for example, with an anti-HTV-1 gene therapeutic)
  • the anti-disease CTLs may be used alone or in combination with a proportion of anti-disease gene-containing CD4+ T- lymphocytes and /or a proportion of anti-disease gene-containing hematopoietic progenitor cells, the latter population being able to produce a proportion of anti-disease gene-containing myeloid (monocyte/macrophages) and lymphoid cells (including CD8+ and CD4+ T-lymphocytes), such that the cell population(s) impact on disease (e.g., HTV- 1 infection and disease) progression.
  • disease e.g., HTV- 1 infection and disease
  • the present invention provides a therapeutic cell product comprising a cytotoxic T-lymphocyte population primed for virus-specific CTL activity produced according to the method of the first aspect.
  • the present invention provides a therapeutic cell product comprising a cytotoxic T-lymphocyte population transduced with virus-inhibiting nucleic acid and primed for virus-specific CTL activity produced according to the method of the second aspect.
  • a method of treating a subject with an infectious disease comprising:
  • nnAPC non-naturally occurring antigen-presenting cells
  • step (c) incubating a population of CD8+ cells obtained from the white blood cells in step (b) with the nnAPC cells;
  • step (e) introducing the CD8+ cells from step (d) into the subject.
  • the subject has more than one infectious disease, and at least one virus-specific antigenic peptide specific for each infectious disease is utilised in step (a).
  • the virus inhibiting nucleic acid utilised in a preferred method of the invention may for instance be specific for a disease selected from the group consisting of Human papilloma virus, Cytomegalovirus, Epstein Barr Virus, Hepatitis A, Hepatitis B,
  • Hepatitis C Hepatitis D
  • Hepatitis E Measles, Mumps, Polio, Rubella, Influenza, Yellow Fever, Japanese Encephalitis, Dengue, Rabies, Rotavirus, Varicella Zoster, Chikungunya Rift Valley Fever, Respiratory Syncitial Virus, Herpes Simplex, Coronaviruses, Marburg, Ebola, California Encephalitis Virus, JC Virus, Lymphocytic Choriomeningitis Virus, Parvovirus, Rhinovirus, Smallpox, HTLV-1, HTLV-2, and HTV.
  • the method of treatment aspect may also comprise incubating a population of CD4+ T lymphocytes obtained from the white blood cells in step (b), and /or a population of CD34+ haematopoietic progenitor cells with the nnAPC cells prior to introducing the CD4+ T lymphocytes and /or CD34+ progenitor cells into the subject.
  • one or more supportive cytokines are added to the
  • CD4+ T lymphocytes and /or CD34+ haematopoietic progenitor cells may be introduced into the population of CD4+ T lymphocytes such that the virus inhibiting nucleic acid is expressed in the lymphocytes, and /or into the population of CD34+ haematopoietic progenitor cells such that the virus inhibiting nucleic acid is expressed in the progenitor cells, prior to the CD4+ T lymphocytes and /or CD34+ progenitor cells being introduced into the subject.
  • treatment interruptions may be used to assist the administered CTL's (alone or in combination with other cell types) to impact on disease.
  • the supportive cytokines added to the CD8+ cells may be selected from the group consisting of IL-2, IL-4, IL-7, IL-15 and IL-21.
  • Figure 1 provides an illustration of the location of a ribozyme target site within the HTV- 1 genome.
  • Panel A provides a schematic diagram of the TTJV-1 genome showing location of replicative, regulatory and accessory genes;
  • Panel B provides the ribozyme sequence together with the complementary target and hybridizing sequence within the tatgene. The target GUA cleavage site is circled; and
  • C provides the location of the GUA target sequence in the genes encoding Tat and Vpr proteins.
  • FIG. 2 provides an illustration of the various major steps of the method of this invention.
  • Figure 3 provides an illustration of the antigen-presenting method involved in Figure 2 above. Apheresis yields peripheral blood mononuclear cells from which CD8+ T- lymphocytes and monocytes are isolated.
  • Figure 4 provides an illustration of the ontogeny of the various cell types - CD8+, CD4+, CD34+.
  • ATM-V The ATM-V medium for cell culture APC Antigen-presenting cells CD8 + CD8 + T cells CD34+ cells cells, which have the CD34+ antigen on their surface; a subset of hematopoietic progenitor cells.
  • DzyNA a method for real-time PCR detection and quantification of DNA or RNA such as that described in U.S. Patent 6,140,055 and U.S. Patent
  • CD95 epitope on T cells HP cell Hematopoietic Progenitor Cell; a pluripotential cell that in vivo continuously gives rise to all of the various lineages of the hematopoietic system.
  • LNL6 a urine retroviral vector, derived from Moloney
  • Murine Leukemia Virus that has the replicative genes deleted and the neomycin phosphotransferase (nee/) gene inserted.
  • the vector is based on the retroviral plasmid, pLNL6, which contains the replication-incompetent retroviral vector LNL6 (Genbank accession number
  • RRz2 retroviral vector consisting of LNL6 with Rz2 inserted into the 3' untranslated region of ned.
  • ex vivo' or "ex vivo therapy” refers to a therapy where biological materials, typically cells, are obtained from a patient or a suitable alternate source, such as, a suitable donor, and are modified, such that the modified cells can be used to treat a pathological condition which will be improved by the long-term or constant delivery of the therapeutic benefit produced by the modified cells.
  • Treatment includes the re-introduction of the modified biological materials, obtained from either the patient or from the alternate source, into the patient.
  • a benefit of ex vivo therapy is the ability to provide the patient the benefit of the treatment, without exposing the patient to undesired collateral effects from the treatment.
  • cytokines are often administered to patients with cancer or viral infections to stimulate expansion of the patient's CTLs.
  • cytokines often cause the onset of influenza like symptoms in the patients.
  • cytokines are used to stimulate expansion of the CTLs outside of the patient's body, and the patient is spared the exposure and the consequent side effects of the cytokines.
  • the subject can be treated concurrently with low level dosages of cytokines, such as, for example, IL-2.
  • the effect of the IL-2 is to enhance antigen specific CTL persistence.
  • MHC major histocompatibility complex
  • HLA human leucocyte antigens
  • epitope refers to a peptide derived from an antigen capable of causing a cellular immune response in a mammal. Such peptides may also be reactive with antibodies from an animal immunized with the peptides. Such peptides may be about five to twenty amino acids in length preferably about eight to fifteen amino acids in length, and most preferably about nine to ten amino acids in length.
  • IL-2 refers to a cytokme which stimulates the immune system and which exerts its biological effects following binding to specific receptors on the surface of target cells.
  • IL-2 has many biological effects, for example, it is known to induce the stimulation of activated B and T cells (including cytotoxic T cells), natural killer (NK) cells, and lymphokine activated killer (LAK) cells.
  • IL-2 may be obtained as a prescription drug, for example, PROLEUKTN®, manufactured by Chiron Corporation (Emeryville, CA).
  • IL-2 may be prepared from various sources and by different methods, as disclosed in numerous U.S. patents.
  • patents include, but are not limited to, the preparation of IL-2 from T cells, such as from hybrid murine T cell lines or malignant human T cell lines, as disclosed in U.S. Pat. No. 4,407,945, 4,473,642, and 4,401,756, respectively, and the preparation of recombinant human IL-2 as disclosed in U.S. Pat. No. 4,992,367, 4, 07,945, and 4,473,642.
  • the present invention is directed at obtaining and preparing the therapeutic dose of an enriched pool of disease-specific CD8+ Cytotoxic T Lymphocytes (CTLs) a proportion of which, in one embodiment, are transduced with a therapeutic gene, for delivery to the patient.
  • the therapeutic gene is preferably a gene or gene construct capable of limiting the dissemination of an infectious disease, and preferably a viral disease.
  • CTLs will be used alone or in combination with gene-containing CD4+ T- lymphocytes and/ or gene-containing CD34+ hematopoietic progenitor cells. While the invention is exemplified with reference to the treatment of HTV, the invention is not limited thereto and methods described herein may be used in the treatment of other viral infections.
  • a preferred outline of the present invention is provided in Figure 2 and 3 and includes apheresis of a patient's blood to obtain peripheral blood mononuclear cells, either with or without G-CSF mobilization (required if CD34+ hematopoietic progenitor cells are required).
  • CD8+, CD4+, and/ or CD34+ T lymphocytes are separately isolated, cultured, optionally transduced, exposed to peptide, cultured, harvested and infused.
  • the invention contemplates that CD8 + cells, CD8+ cells in combination with CD4+ cells, CD8+ cells in combination with CD34+ cells or a combination of all three cell types can be infused back to the patient.
  • non-naturally occurring antigen-presenting cells are prepared.
  • the nnAPC are capable of being loaded simultaneously with at least one and up to fifteen different exogenous peptides where each peptide is preferably at least eight and more preferably eight to ten amino acids in length, and are capable of presenting the peptide molecules on the surface of their cells.
  • the peptides used to load the antigen presenting cells are associated with infectious diseases and preferably are associated with viral disease.
  • a variety of peptides can be used, but preferably the peptides are known to stimulate cytotoxic T cell responses.
  • Peptides, including viral-derived peptides, that can stimulate cytotoxic responses have been identified in the art.
  • methods and assays that are known to determine which peptide fragments can stimulate cytotoxic T cell responses and can therefore be used to identify other peptides that can be used in this invention.
  • the examples employ exemplary peptides derived from HTN protein to stimulate an HlV-specific cytotoxic T cell response. Cytotoxic T lymphocytes (CTLs) are one of the natural barriers to viral infection.
  • CTLs Cytotoxic T lymphocytes
  • HTV Human Immunodeficiency Virus
  • one of the major obstacles to clearing infection is that the virus decreases the production of the CTLs, in part by infecting and destroying this cell population.
  • Methods described herein can provide a population of disease-specific CTLs with or without gene modification.
  • the gene modification should mean that the disease-specific CTLs are not subject to disease induced depletion, thereby acting to ameliorate disease.
  • the CTL population (with or without gene modification thereof) can be used alone or in combination with other gene- modified cells such as CD4+ T lymphocytes or CD34+ hematopoietic progenitor cells.
  • the CD8+ T-lymphocytes are first incubated with disease-specific peptide epitopes in the presence of rvnAPCs and cultured for a further period of time.
  • the CD8 + cells are incubated with the nnAPC cell line for about six to seven days.
  • Supportive cytokines such as Interleukin-2 (IL-2) and Interleukin-7 (IL-7) can be added to the media 4 to 5 days after stimulation with the nnAPCs.
  • IL-2 Interleukin-2
  • IL-7 Interleukin-7
  • 20U/ml IL-2 and 30 U/ml of IL-7 are added to the media.
  • the nnAPC that express the peptides are further modified to express other peptides or polypeptides that can enhance the treatment of the subject.
  • the nnAPC can express polypeptides associated with accessory molecules such as, lymphocyte function antigens (LFA-1, LFA-2 and LFA-3), intercellular adhesion molecule 1 (ICAM-1), and /or T-cell co-stimulatory factors (CD2, CD28, B7), in order to enhance cell-cell adhesion or transduce additional cell activation signals.
  • LFA-1, LFA-2 and LFA-3 intercellular adhesion molecule 1
  • CD2, CD28, B7 T-cell co-stimulatory factors
  • the CD8+ cell population is transduced with an infectious disease protective gene therapeutic.
  • the infectious disease protective gene therapeutic is introduced such that it is expressed in not only the transduced cells, but also the progeny of the transduced cells.
  • the infectious disease protective gene therapeutic comprises nucleic acid and methods for introducing nucleic acid in a variety of forms into cells and is well known in the art.
  • the infectious disease protective gene therapeutic is directed to viral disease.
  • the therapeutic is a ribozyme.
  • CD8+ CTLs are transduced with an anti-HIV gene construct, RRz2, to produce a therapeutic effect by decreasing viral load and increasing CD4+ cell count as a result of the HTV directed CTL activity of the cells containing the therapeutic gene.
  • RRz2 an anti-HIV gene construct
  • HTV-1 novel anti-Human Immunodeficiency Virus 1
  • HTV-1 novel anti-Human Immunodeficiency Virus 1
  • These approaches include intracellular expression of transdominant proteins (Smythe et al. 1994), intracellular antibodies (Marasco et al. 1998), antisense ribonucleic acid (RNA) (Sczakiel et al. 1991), viral decoys (Kohn et al. 1999), and catalytic ribozymes (Sarver et al. 1990; Sun et al. 1996).
  • the CD8+- derived, CTL enriched population is further incubated with the same disease-specific peptide epitopes in the presence of an adherent population of autologous monocytes which were produced following the isolation of the adherent cell population from the peripheral blood mononuclear cells.
  • the CD8 + cells are incubated with peptide-loaded adherent cells for about six to seven days by: mixing adherent cells - obtained from the peripheral blood mononuclear cell population, most preferably adherent monocytes from the CD8+ depleted peripheral blood collected from said subject or a suitable donor with about 10 to 50 ⁇ g/ml of each peptide of interest.
  • Peripheral blood monocytes are isolated from the peripheral blood mononuclear cell population following apheresis.
  • the peripheral blood monocyte suspension is preferably irradiated with a sufficient dose of ⁇ -radiation necessary (to prevent further cell proliferation of the PBMCs while maintaining their stimulation capacity) such as a dose in the range of about 3,000 to 7,000 rads, preferably about 5,000 rads.
  • Adherent peripheral blood monocytes are isolated from the cell suspension. These adherent cells are loaded with peptide by mixing the cells with about lOng/ml to lO ⁇ g/ml of each peptide.
  • the CD8 + cells are combined with the adherent peripheral blood monocytes at a ratio of about ten CD8 + cells to one peripheral blood monocyte.
  • the cells are incubated for about six to seven days.
  • the combined cells can be further incubated with IL-2 and IL-7 in media.
  • CD8+ cells it is possible to stimulate the suspension of CD8+ cells in a non-specific manner such as by contacting the cells with a mAb directed against the CD3 receptor in the presence of -irradiated "feeder cells” composed of peripheral blood mononuclear cells (PBMCs) or alternatively, CD8-depleted PBMCs using a dose in the range of about 3,000 to 4,000 rads, preferably about 3,500 rads to irradiate the feeder cells.
  • feeder cells composed of peripheral blood mononuclear cells (PBMCs) or alternatively, CD8-depleted PBMCs using a dose in the range of about 3,000 to 4,000 rads, preferably about 3,500 rads to irradiate the feeder cells.
  • the CTL population can be expanded further in the presence of growth factors.
  • the CD8 + suspension can be assayed for suitable CTL activity as well as for CTL purity, sterility and endotoxin content.
  • the cells can be introduced into a subject.
  • Methods for introducing or reintroducing cells into a subject are known in the art, such as those methods that are used to repopulate bone marrow as part of a bone transplantation procedure or those methods used in the clinical trial using CD34+ cells (see Amado, et al. (1999). Human Gene Therapy, 10:2255- 2270).
  • a dose of cells of between 6 - 10 x 10 9 cells are used per treatment and the treatment may be repeated between 1 and 6 times.
  • CTLs are dependent on interleukin-2 for survival and growth. Therefore, in one embodiment, the subjects will have interleukin-2 administered to them prior to or alternatively after the administration of the CTL cell population. Interleukin-2 will likely increase the survival, replication and activity of the CTLs and can also increase survival of other lymphocytic cell types that may be introduced into the subject. In a preferred embodiment, the subject will receive 3 MTU of IL-2 before and/ or after administration of the cell population.
  • IL-2 has been used clinically for both renal cell carcinoma and malignant melanoma.
  • the combination of IFN- -2b and IL-2 in low dose subcutaneous regimens has been described in other clinical settings (Pectasides et al.. Oncology (1998) 55:10-15/ Piga et al., Cancer Immumol Immunotherapy -(1997) 44:348- 35
  • the CD8+ cells produced according to the methods of the present invention are introduced to a subject in combination with CD4+ T-lymphocytes for the treatment of infectious disease.
  • the CTL population will be produced in the same way as in the first aspect and will be processed concurrently with, though at all times subsequent to separation, separately from, the CD4+ population.
  • the CD4+ population is preferably also transduced with the same infectious disease protective gene therapeutic as that with which the CD8+ cells were transduced.
  • the CD8+ cells produced according to the methods of the present invention are introduced to a subject in combination with CD34+ hematopoietic progenitor cells for the treatment of infectious disease.
  • the CTL population will be produced in the same way as in the first aspect and will be processed concurrently with, though at all times subsequent to separation, separately from, the CD34+ HP cell population.
  • the CD34+ HP population is preferably also transduced with the same infectious disease protective gene therapeutic as that with which the CD8+ cells were transduced.
  • the administration of the cells is combined with the use of antiretroviral treatment withdrawals (i.e., treatment interruptions).
  • antiretroviral treatment interruptions refers to the cessation of non-cell therapy antiviral treatment for a period of time and during the non-cell therapy antiviral treatment interruption the methods of this invention are employed or continued.
  • the methods of this invention are useful for infectious diseases of the hematopoietic system generally.
  • the invention is directed towards therapy for HTV, wherein the harvested cells from a HTV positive subject are enriched for the various cell types and the therapeutic gene(s) encodes an anti-HIV product(s).
  • Example 1 The use of non-naturally occurring antigen-presenting cell (nnAPC) derived from Drosophila melanogaster cells for the generation of HTV-specific CTLs
  • the non-naturally occurring antigen-presenting cell are produced from Drosophila melanogaster cells that were modified to express human class I HLA binding and co-stimulatory molecules according to the methods disclosed in PCT publication PCT/US02/ 005748. These cells are capable of presenting up to fifteen different peptide molecules, preferably peptide molecules that are simultaneously exogenously loaded onto the surface of the previously transfected Drosophila cells.
  • a Drosophila cell line which may be used in this way, can be produced as follows:
  • the Schneider S2 cell line was prepared from Drosophila melanogaster (Oregon-R) eggs according to published procedures and has been deposited with the American Type Culture Collection as CRL 10974. S2 cells are grown in commercial Schneider's Drosophila medium supplemented with 10% fetal bovine serum.
  • the pRmHa-3 plasmid vector for expressing MHC Class I and co-stimulatory proteins in S2 cells was derived from the pRmHa-1 expression vector as described in the literature.
  • the vector contains a metallothionein promoter, metal response consensus sequences and an alcohol dehydrogenase gene bearing a polyadenylation signal isolated from Drosophila melanogaster.
  • the plasmid vector pRmHa-3 plasmid is modified to include complementary DNA sequences for human class I HLA A2.1, B7.1, B7.2, ICAM- 1, ⁇ -2 microglobulin and LFA-3, wherein A2.1 can be substituted with any human class I DNA sequence.
  • HLA-A2.1 and ⁇ -2 microglobulin Reverse transcription-PCR from K562 cells using primers derived from the published sequence
  • LFA-3 Reverse transcription-PCR from HL-60 cells (ATCC CCL-240) using primers derived from the published sequence
  • the S2 cells are transfected with a phsneo plasmid and said pRmHa-3 plasmid containing complementary DNA.
  • Stably transfected cells are selected by culturing in Schneider's medium containing geneticin. Twenty-four hours before use expression of the transfected genes was induced by the addition of CuS0 4 .
  • insect growth media are commercially available from a number of vendors, such as, SchneiderTM's Drosophila Medium, Grace's Insect Media, and TC-100 Insect Media.
  • insect growth media can be prepared by one of ordinary skill in the art.
  • the media will include components necessary to promote and sustain the growth of insect cells, such as, inorganic salts (for example, calcium chloride, magnesium sulfate, potassium chloride, potassium phosphate, sodium bicarbonate, sodium chloride, and sodium phosphate), amino acids, various carbohydrate and chemical species (Schneider, Imogene (1964) Exp. Zool. 156:1:91).
  • the media can also include vitamins, minerals, and other components that aid in the growth of insect cells.
  • This Drosophila cell line is used to make nnAPCs as follows:
  • the Drosophila cell line is loaded with at least one and up to fifteen different exogenous peptides where each peptide is preferably eight to ten amino acids in length.
  • the peptides are peptide molecules containing immunostimulatory epitopes, preferably epitopes that can be demonstrated in vitro to stimulate CTL activity.
  • the epitopes are preferably identified from infectious agents, preferably viruses and more preferably, an immunodeficiency virus, such as, HTV.
  • HLA-A2 T cell epitopes for HTV (ILKEPVHGV HTVpol and SLYNTNATL HTVgag). These two epitopes were validated by generating specific CTLs following stimulation with nnAPCs loaded with those two peptides.
  • HTV epitopes that may be used are as found in Part IIA "HTV CTL Epitope Tables”; Part TIB “HTV CTL Epitope Maps”; Part IIC “References” of HTV Molecular Immunology 2001 (Eds Korber et al) Division of AIDS, ⁇ IAID http:/ / hiv-web.Ianl.gov/ immunology.
  • CD8 + cells are isolated from leukapheresis samples by positive selection using the DynabeadsTM isolation procedure (Dynal).
  • An anti-human CD8 mouse monoclonal antibody (50 ⁇ g/ml in human gamma globulin [Gammagard ® ]) is added to washed cells in Dulbecco's PBS supplemented with 1% human serum albumin (Baxter-Hyland) and 0.2% Na citrate.
  • the cells After incubation at 4°C for forty-five minutes with gentle mixing, the cells are washed and re-suspended in the same buffer containing Dynal magnetic beads (DynabeadsTM) coated with sheep anti-mouse IgG at a bead to cell ratio of 1:1. The cells and beads are placed into a sterile tube and gently mixed at 4°C for forty-five minutes. At the end of this time, the antibody-bound cells are removed magnetically using the MPC-1 ® separator according to the manufacturer's instructions (Dynal). Dissociation of the CD8 cell-bead complex is achieved by incubation at 37°C for forty-five minutes in the presence of CD8 peptide 59 . 70 (AAEGLDTQRFSG; SEQ ID NO: 12). Free beads are removed magnetically and the CD8 cells are counted and analyzed by flow cytometry to evaluate purity.
  • Dynal magnetic beads Dynal magnetic beads coated with sheep anti-mouse IgG at a bead to
  • Example 3 Purification and Sensitization of CD8+ Cells Transfected Drosophila S2 cells are incubated in Schneider's medium (10 6 cells /ml) supplemented with 10% fetal calf serum and CuSO 4 at 27°C for twenty-four hours. Cells are harvested, washed and re-suspended in Insect X-press medium
  • the S2 cells are mixed with CD8 + cells at a ratio of 1:10 in RPMI medium (Gibco) supplemented with 10% autologous serum.
  • the cell mixture is incubated for preferably at least four days and more preferably between 6-7 days at 37°C during which the Drosophila cells die off.
  • IL-2 (20 U/ml) and IL-7 (30 U/ml) are added to selectively expand the HTV-specific CTL population.
  • the CTL enriched CD8+ T-lymphocytes are transduced with an infectious disease protective gene therapeutic, for example, a ribozyme conferring protection to HTV, such as RRz2.
  • an infectious disease protective gene therapeutic for example, a ribozyme conferring protection to HTV, such as RRz2.
  • Such transduction is carried out, for example, by means of a retrovirus, such as, LNL6 to involve the introduction of the genetic agent, such that it is expressed in the cells transduced and in their subsequent progeny cells (e.g., Knop etal (1999).
  • Adherent monocytes are loaded with the peptide by incubation for 90 minutes in Hepes- buffered RPMI medium containing 10% autologous serum and lOng/ml-10 ⁇ g/ml of one or more HTV-stimulatory peptides such as HTV (ILKEPVHGV HTVpol and SLYNTVATL HTVgag).
  • HTV HTV-stimulatory peptides
  • Other examples of HTV epitopes that may be used are as found in Part IIA "HTV CTL Epitope Tables"; Part ITB "HTV CTL Epitope Maps"; Part IIC
  • effector cells are non-specifically expanded by culturing them in RPMI medium supplemented with autologous serum, anti-CD3 monoclonal antibody (OKT ® 3), and IL-2 and ⁇ irradiated autologous PBMCs (such as a dose in the range of about 3,000 to 4,000 rads, preferably about 3,500 rads) or alternatively, CD8-depleted PBMCs alone or IL-2 and ⁇ irradiated autologous PBMCs (such as a dose in the range of about 3,000 to 4,000 rads, preferably about 3,500 rads) alone.
  • RPMI medium supplemented with autologous serum, anti-CD3 monoclonal antibody (OKT ® 3), and IL-2 and ⁇ irradiated autologous PBMCs (such as a dose in the range of about 3,000 to 4,000 rads, preferably about 3,500 rads) alone.
  • autologous serum such as a dose in the range of
  • Malme 3M cells (ATCC) are used as target cells in a 51 Cr release assay. 5 x 10 6 Malme 3M cells in RPMI medium containing 4% fetal calf serum, 1% HEPES buffer and 0.25% gentamycin are labeled at 37°C for one hour with 0.1 mCi 51 Cr. Cells are washed four times and diluted to 10 5 cells /ml in RPMI with 10% fetal bovine serum (HyClone). In a 96-well microtiter plate, 100 ⁇ l effector CTLs and 100 ⁇ l peptide-loaded, 51 Cr-labeled Malme 3M target cells are combined at ratios of 100:1, 20:1 and 4:1 (effector: target).
  • K562 cells are added at a ratio of 20:1 (K562:Malme 3M) to reduce natural killer cell background lysis.
  • Non-specific lysis is assessed using the non-tumor HLA-A2.1 fibroblast cell line, Malme 3. Controls to measure spontaneous release and maximum release of 51 Cr are included in duplicate. After incubation at 37°C for six hours, the plates are centrifuged and the supernatants counted to measure 51 Cr release.
  • Percent specific lysis is calculated using the following equation:
  • CD8 + cells before and after in vitro activation are analyzed for a number of cell surface markers using fluorescent monoclonal antibodies and FACS analysis.
  • Infectious agent-specific CD8+ derived CTLs can also be used in combination with CD4+ T-lymphocytes for the treatment of infectious disease.
  • the CTL population is produced as described above and is processed concurrently with, though at all times subsequent to separation, separately from, the CD4+ population.
  • the CD4+ population is prepared and used as follows.
  • the CD4+ cells are harvested from a subject, subsequent to the isolation of CD8+ cells from the apheresis product.
  • CD4+ cells are transduced with retrovirus, activated and introduced into tissue culture.
  • CD4+ cells are stimulated with materials such as OKT3 and IL-2.
  • the cells are transduced with the anti-infectious disease therapeutic at day 3-5 of culture.
  • the CD4+ cells can also be further stimulated and expanded using IL-2 and appropriate medium changes.
  • the CD4+ cells are harvested, washed, checked for sterility and infused into the patient.
  • the CD4 + enriched T-lymphocytes were then incubated in 15 ml of either control (LNL6) or ribozyme (RRz2) GMP-grade virus (in duplicate) for 15-30 minutes at 37°C and stimulated with OKT3 (anti-CD3) antibody (Orthoclone; Janssen-Cilag, Belgium) at a concentration of 50ng per 10 7 cells.
  • the enriched CD4 + T-lymphocytes were then introduced into four pre-cultured
  • Peripheral Blood Lymphocyte-MPS artificial capillary cartridges (Cellco CellmaxTM, USA) by syringing the 15ml cell suspension into a cartridge side port.
  • the cells were cultured initially in AIM-V medium (Gibco BRL) containing 5% heat inactivated autologous plasma.
  • AIM-V medium Gibco BRL
  • IL-2 rIL-2; Chiron, USA
  • the proportion of plasma was reduced stepwise by using non-plasma containing media for subsequent media additions, and cultures were almost plasma free by day 7 (less than 0.1% remaining).
  • media additions to the reservoirs were AIM- V only.
  • the concentration of L-lactate in the module reservoir was determined by taking a sample from the reservoir arid measuring the L- lactate concentration (L-lactate reagent Sigma, USA). This determination was based on the approximation that 4 x 10 6 PBL produces 1 mg lactate per day (Cellco). The medium in the reservoir was changed (in order to add fresh growth additives and remove metabolites) when the concentration of lactate exceeded 0.6 mg/ml.
  • the second retroviral transduction was conducted when the target CD4 + lymphocytes reached exponential growth phase (generally days 3-5 of culture) as monitored by lactate concentration. This was conducted as two separate transductions performed on consecutive days (30 ml VCM was added in the first transduction and 70 ml VCM in the second). Transduction was effected by direct injection of VCM through the cartridge sideports as previously described (Knop et al, 1999). Prior to injection into the sideport, 100 U/ml IL-2 and protamine sulphate (Fisons/ Rhone-Poulenc Rorer, USA) were added to the VCM and protamine sulphate also added to the culture reservoir. The concentration of protamine sulphate added yielded a final concentration of 5 ⁇ g/ml.
  • Cells were harvested when cultures had reached plateau growth phase (generally day 10 of culture), as determined by no further increase in L-lactate production over two consecutive days.
  • the cells were harvested, according to the Manufacturer's instructions, by injecting media into the extra-capillary space through the capillary pores and flushing the cartridges with medium. This was followed by three cell washes (centrifugation at 200g for 5- minutes) then resuspension in an infusion buffer containing 0.18% saline, 4% glucose (Baxter) and 2.5% human serum albumin (Albumex 20,Red Cross Blood Bank, Australia). Samples were removed at this time for further testing: sterility (aerobic and anaerobic culture), analysis by flow cytometry and archival storage.
  • the present invention also provides for the use of infectious agent-specific CD8+ derived CTLs in combination with CD34+ HP cells (also termed HP cells) for the treatment of infectious disease.
  • CD34+ HP cells also termed HP cells
  • the CTL population will be produced in the same way as in the first aspect and will be processed concurrently with, (though at all times subsequent to separation) separately from the CD34+ population.
  • the CD34+ population can be prepared and used for example by obtaining from said subject a population of viable cells comprising hematopoietic progenitor (HP) cells of the CD34+ class, treating and/ or culturing said population of cells to provide a pool of cells comprising a portion of HP cells in excess of 40%, and introducing said therapeutic gene(s) into at least a portion of the HP cells within said pool of cells such that said therapeutic gene(s) is/ are capable of being expressed in said portion of HP cells wherein a resultant pool of viable cells is prepared which includes sufficient HP cells including the therapeutic gene(s) such that, upon delivery to said subject, the subject receives a dose of at least 0.5xl0 6 CD34+ HP cells containing the therapeutic gene(s) per kg body weight.
  • HP hematopoietic progenitor
  • the resultant pool of viable cells are transduced with a disease-treating gene therapeutic and the viable cell pool comprises sufficient therapeutic gene(s) containing CD34+ HP cells such that, upon delivery to said subject, the subject receives a dose of at least 5xl0 6 , more preferably in excess of 2xl0 7 , and even more preferably in excess of 5xl0 7 therapeutic gene(s) containing CD34+ HP cells /kg body weight.
  • the resultant pool of viable cells is such that, upon delivery to said subject, the subject receives a total number of cells (i.e. the HP cells containing the therapeutic gene(s) and all other cells present in the resultant pool of cells) of at least lxl0 7 /kg body weight up to 4xl0 7 cells/kg or more preferably up to 10xl0 7 kg or more).
  • a total number of cells i.e. the HP cells containing the therapeutic gene(s) and all other cells present in the resultant pool of cells
  • the population of cells "harvested" from the subject may be obtained by any of the methods well known in the art.
  • the subject may be treated so as to mobilize HP cells from bone marrow into the peripheral blood (e.g. by administering a suitable amount of the cytokine granulocyte - colony stimulating factor, G-CSF) followed by apheresis filtration.
  • HP cells may be aspirated from bone marrow or cord blood in accordance with well-known techniques.
  • Treatment of the harvested population of cells may include one or more washing steps (e.g. using centrifugation or automated cell washers) and /or de-bulking steps (i.e. to remove excess red blood cells, granulocytes, platelets, T-lymphocytes and), by use of a device such as the Dendreon DACS System (Charter Medical, Winston Salem, NC) and, preferably, a HP cell selection step.
  • HP cell selection ma be achieved by immune affinity or flow cytometry techniques.
  • the HP cell selection step selects CD34+ cells or in another embodiment may involve antigen depletion of mature/ committed hematopoietic cells, thereby enriching for HP cells.
  • Such cells can be selected using a variety of selection devices such as, but not limited to, the Nexell/ Baxter Isolex 3001 (Irvine, CA), the Miltenyi CliniMACS,(Miltenyi; Biotech GmBH, Bergisch Gladbach, Germany), Stem Cell Technologies (Vancouver, BC, Canada) StemSep Device.
  • the treatment of the harvested population of cells may also involve a cell culturing step to increase cell numbers and especially to increase the number of selected HP cells.
  • Cell culturing is also required to introduce the therapeutic gene(s) and cell culturing may additionally be conducted after introduction of the therapeutic gene(s) into at least a portion of the HP cells to expand the number of such gene(s)-containing HP cells.
  • the initial treatment steps results in the obtaining of, and enriching for, HP cells.
  • the definition of the percentage of HP cells requires a measurable aspect of these cells such as CD34 antigen positivity. It is to be understood that the treated pool of cells comprises at least 40%, more preferably at least 60% and most preferably at least 80%, HP cells.
  • the therapeutic gene(s) or nucleic acid sequence(s) into at least a portion of the HP cells may be achieved with any of the methods well known to the art, but most conveniently through transduction using retroviral vectors or other viral or non-viral (DNA or RNA) vectors carrying the therapeutic gene(s) or nucleic acid sequence(s), and, preferably, a transduction-facilitating agent (e.g. for retroviral vectors, the CH296 fragment of fibronectin known as RetroNectin).
  • the HP cells containing the therapeutic gene(s) or nucleic acid sequence(s), and cells derived therefrom i.e. from subsequent lymphoid and myeloid hematopoiesis, contain and are capable of expressing the therapeutic gene(s) or nucleic acid sequence(s).
  • infusion is performed at day 3 post introduction to cell culture.
  • the therapeutic gene(s) may encode a product selected from proteins (e.g. transdominant proteins and intracellular antibodies), antisense molecules (eg antisense RNA), RNA decoys, aptamers, interfering RNA and catalytic ribozymes.
  • proteins e.g. transdominant proteins and intracellular antibodies
  • antisense molecules e.g antisense RNA
  • RNA decoys e.g antisense RNA
  • aptamers e.g. interfering RNA and catalytic ribozymes.
  • the first step of this procedure uses an agent to mobilize HP cells from the bone marrow into the peripheral blood.
  • An example here is the use of Granulocyte Colony Stimulating Factor (G-CSF, NeupogenTM), which is administered to the patient subcutaneously, at least at 10 ⁇ g/kg/day and preferably at 30 ⁇ g/kg/day, once daily, for up to five consecutive days.
  • G-CSF Granulocyte Colony Stimulating Factor
  • CBCs Complete Blood Counts
  • differential and platelet count are performed daily during G-CSF administration to assess the extent of the leucocytosis.
  • a blood sample for CD34+ cell count is drawn on day 3 of G-CSF administration to ensure that the peripheral blood CD34+ count is greater than 20 cells/mm 3 prior to the start of apheresis. Failure to attain this CD34+ cell number does not however prevent apheresis on days 5 and 6 of G-CSF administration.
  • Apheresis is a method of "blood filtration” to obtain the mononuclear cell fraction of the peripheral blood. It is conducted with a Cobe Spectra (Gambra), Hemonetics (Domedica) or Amicus (Baxter) machines on at least two separate occasions, (preferably on days 5&6 following mobilization, where day 1 is the first day of induced mobilization), though in other examples this can be done on earlier or later days by determining the day at which the peripheral blood CD34+ count is greater than 5 cells /mm 3 or more preferably 10 cells /mm 3 and most preferably 20 cells /mm 3 .
  • this apheresis yields cellular product from about 5 Liters (L) of blood flow, though preferably this will be 5-10 L, but more preferably 10-20 L, and more preferably still 20L or greater.
  • Product from each apheresis is either treated separately or, in a preferred embodiment, pooled after the second apheresis.
  • Total cell counts, and absolute CD34+ cell numbers are recorded. Use of these aphereses will produce up to greater than 5xl0 6 , preferably greater than 2xl0 7 , more preferably greater than 4xl0 7 HP (as measured by CD34 positivity) cells /kg
  • the pooled cells are washed. This is done by cell centrifugation or more preferably using an automated cell washer, in one example this cell washing is done by using a Nexell CytoMate washer.
  • the cells from the apheresis procedure(s) are "de-bulked" - using a system like a Charter Medical DACS-SCTM system.
  • the two apheresis products are de-bulked on the day of collection and the first product stored until the second product has been de-bulked.
  • CD34+ cells are selected from the post-washing product by using the Isolex 300i, Miltenyi or a lineage depletion strategy of cells expressing markers (e.g. CD2, CD3, CD14, CD16, CD19, CD24, CD56, CD66b glycoprotein A, StemSep).
  • the enriched pool of CD34+ or lineage depleted cells preferably comprises at least 40%, more preferably at least 60% and most preferably at least 80% cells of this type.
  • the cells are washed by centrifugation or by using the Nexell CytoMate or similar.
  • the cells are counted and placed at preferably lxlO 5 to 5xl0 6 cells/ml into cell culture flasks, cell culture bags or in a preferred embodiment into 1,000ml (390cm 2 ) Nexell Lifecell X-Fold Culture Bag or similar with Iscove's Modified Dulbecco's Medium plus 10% Fetal Bovine Serum (FBS) containing cytokines /growth factors.
  • this cytokine/ growth factor mixture consists of Stem Cell Factor (50ng/ml) and Megakaryocyte Growth and Development Factor lOOng/ml). Steps 3-9 will result in up to 12xl0 7 HP cells or more (as assessed by CD34 positivity) per kg.
  • the cells are harvested from the first flask, tissue culture bag, including a preferred embodiment of a Lifecell Culture Bag or similar and using the Cytomate device or similar, resuspended in retroviral supernatant (an example of this is a 200 ml aliquot) and transferred into a second tissue culture container, one type of which is the Lifecell X-Fold Culture Bag which have a retrovirus transduction facilitating agent.
  • retroviral supernatant an example of this is a 200 ml aliquot
  • Such agents include polybrene, protamine sulphate, cationic lipids or in a preferred embodiment, in a tissue culture container that has been pre-coated with RetroNectin at l-4mcg/ cm 2 .
  • the transfer procedure will be repeated using the CytoMate or similar; for this second transduction cells are either transferred to a new tissue culture container (polybrene, protamine sulphate) or returned to the same or similar RetroNectin-coated container from which they came. In a preferred embodiment, this is done in a fresh aliquot of retroviral supernatant and cultured overnight. In other embodiments, this is either not done or repeated several times for similar periods of time. An aliquot of the retroviral supernatant(s) is collected for sterility testing. This will result in up to 6xl0 7 gene-containing HP cells or more (as assessed by CD34 positivity) per kg.
  • This number is determined by quantitative assay such as DzyNA PCR.
  • the transduction efficiency will be at least 20%, and preferably in the range from 30-50%, and more preferably greater than 50%.
  • cells are harvested and washed using standard cell centrifuge or automated systems such as the Cytomate samples of cell culture. This will yield up to 5.7 x 10 7 gene-containing HP cells or more (as assessed by CD34 positivity) per kg.
  • Cells are resuspended in a physiologic infusion buffer containing 5% human serum albumin or similar as carrier. Aliquot samples are removed for sterility (aerobic, anaerobic, fungal, mycoplasma). Infusion product is not released until the results of endotoxin (LAL) and gram stain testing are available.
  • LAL endotoxin
  • the CD34+ cell preparation is administered to the patient pre-medicated as appropriate.
  • the patient receives a single infusion of 0.5-6 x 10 7 transduced CD34+ cells per kilogram of body weight (cells/ kg) in the physiologic infusion buffer containing 5% human serum albumin or similar as carrier.
  • the dose of transduced CD34+ cells per patient will depend on the efficiency of each step of the mobilization, apheresis, isolation, culture and transduction procedures.
  • the total number of CD34+ cells (transduced and non- transduced) is determined by cell counting and flow cytometry.
  • the introduced gene-containing HP cells give rise to a chimeric hematopoietic system in which there is a percentage of gene-containing HP cells in the bone marrow.
  • this percentage of gene-containing HP cells is at least 5%, preferably greater than 10% and more preferably greater than 20%.
  • the subject does not require myeloablation of the bone marrow or other marrow conditioning regimen, and the step of delivering the cells results in the subject receiving a dose of at least 0.5xl0 6 CD34+ cells containing the therapeutic gene(s)/kg body weight.
  • the step of delivering the cells results in the subject receiving a dose of at least 0.5xl0 6 CD34+ cells containing the therapeutic gene(s)/kg body weight.
  • this is a "sufficient" dose of cells to produce a chimeric hematopoietic system that will yield persistence (presence of gene-containing cells for greater than one year post- infusion) of anti-HTV product-containing mature lymphoid (CD4+ and CD8+ T- lymphocytes) and myeloid (monocyte/macrophages) cells.
  • Example 8 Use of IL— 2
  • IL-2 is used post infusion to benefit treatment.
  • a minimum dose of IL-2 contemplated for use is 2-4 MIU/m2 (million international units per meter squared), subcutaneously for 12 days every 3 weeks and preferably 3 MIU/m 2 .
  • Immune activation is monitored and significant increases in lymphocytes, activated CD4+ and CD8+ T cells, NK cells, and monocyte DR expression are monitored.
  • the presence of the high affinity IL-2 receptor (CD25/CD122/CD132) on the surface of the CD8+ cells suggests that these cells are capable of responding to low doses of IL-2 in vivo.
  • the cells are delivered to the subject in accordance with routine methods such as cell infusion.
  • the cells are preferably delivered with a pharmacologically-acceptable carrier (e.g. 5% Human Serum Albumin).
  • a pharmacologically-acceptable carrier e.g. 5% Human Serum Albumin.
  • the subject may or may not be first (ie before re-infusion of the cells) subjected to myeloablation of the bone marrow or other hematopoietic conditioning regimens.
  • DzyNA-PCR quantitative real time PCR methodology
  • DzyNA-PCR Todd et al. 2000
  • Patent Nos. 6,140,055 and 6,201,113 provide a general strategy for the detection of specific genetic sequences associated with disease or the presence of foreign agents.
  • the method provides a system that allows homogeneous nucleic acid amplification coupled with real-time fluorescent detection in a single closed vessel.
  • the strategy involves in vitro amplification of genetic sequences using a DzyNA primer which harbors the complementary (antisense) sequence of a 10:23 DNAzyme (Santoro et al. 1997).
  • amplicons are produced which contain active (sense) copies of DNAzymes that cleave a reporter substrate included in the reaction mix.
  • the accumulation of amplicons during PCR is monitored by changes in fluorescence produced by separation of fluoro/ quencher dye molecules incorporated into opposite sides of a DNAzyme cleavage site within the reporter substrate. Cleavage of this reporter substrate indicates successful amplification of the target nucleic acid sequence.
  • Real-time measurements can be performed on the ABI PRISM® 7700 Sequence Detection System (Applied Biosystems) or other thermocyclers that have the capacity to monitor fluorescence in real time.
  • DzyNA-PCR is a general strategy for the detection of specific genetic sequences associated with disease or the presence of foreign agents.
  • the method provides a system that allows homogeneous nucleic acid amplification coupled with real time fluorescent detection in a single closed vessel.
  • the strategy involves in vitro amplification of genetic sequences using a DzyNA primer which harbors the complementary (antisense) sequence of a 10:23 DNAzyme.
  • amplicons are produced which contain active (sense) copies of DNAzymes that cleave a reporter substrate included in the reaction mix.
  • the accumulation of amplicons during PCR is monitored by changes in fluorescence produced by separation of fluoro/ quencher dye molecules incorporated into opposite sides of a DNAzyme cleavage site within the reporter substrate.
  • Cleavage of this reporter substrate indicates successful amplification of the target nucleic acid sequence.
  • Real time measurements can be performed on the ABI Prism 7700 Sequence Detection System or other thermocyclers that have the capacity to monitor fluorescence in real time (eg Corbett Rotor-Gene, Stratagene Mx 4000, Cepheid SmartCycler, Roche LightCycler, Biorad iCycler etc).
  • DzyNA PCR protocols have been developed for analysis of vectors and therapeutic agents that contain the neomycin resistance gene. This assay has various uses including estimation of the percent transduction of cells and monitoring the presence and quantification of transduced cells, or their progeny, within patients undergoing gene therapy.
  • the reporter substrate, Sub G5-FD was synthesised by Trilink Biotechnologies (California, USA).
  • Sub G5-FD (illustrated below) is a chimeric molecule containing both RNA (shown below in lower case) and DNA nucleotides. It has a 3' phosphate group that prevents its extension by DNA polymerase during PCR.
  • Sub G5-FD was synthesised with FAM (F) and DABCYL (D) moieties attached to the "T" deoxyribonucleotides indicated. The cleavage of the reporter substrate can be monitored at 530nm (FAM emission wavelength) with excitation at 485nm (FAM excitation wavelength).
  • the 5' PCR primer (5L1A) hybridizes to the neomycin resistance gene.
  • the 3' primer (3LlDz5) is a DzyNA PCR primer, which contains (a) a 5' region containing the catalytically inactive antisense sequence of an active DNAzyme and (b) a 3' region, which is complementary to the neomycin resistance gene.
  • the amplicons produced by extension of 5L1A contain both neomycin resistance sequences and catalytically active sense copies of a DNAzyme incorporated in their 3' regions.
  • the active DNAzyme is designed to cleave the RNA/DNA reporter substrate Sub G5-FD.
  • the human cell line CEMT4 was obtained from the American Type Culture Collection (Rockville, MD). CEMT4 cells were transduced with retrovirus containing the neomycin resistance gene. Genomic DNA was isolated from CEM T4 cells, as well as CEMT4 cells transduced with retrovirus harboring the Neomycin resistance gene, using the QIAGEN DNeasy Tissue Kit (QIAGEN Pty Ltd, Victoria, Australia. Cat # 69504). DNA extracted from transduced cells was mixed with DNA from untransduced cells (by weight) to obtain the following percentage of transduced DNA -100%, 11%, 1.2%, 0.1%, 0.02% and 0% (ie 100% untransduced CEMT4).
  • Prism 7700 Sequence Detection System denatured at 95°C for 10 minutes, subjected to 10 cycles of 70°C for 1 minute with a temperature decrease of 1°C per cycle, and 94°C for 1 minute. This was followed by a further 60 cycles at 60°C for 1 minute and 94°C for 30 seconds. Fluorescence was measured by the ABI Prism 7700 Sequence Detection System during the annealing /extension phase of the PCR.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biotechnology (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pulmonology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne la production d'une population de lymphocytes T cytotoxiques spécifiques d'un virus, ladite population contenant une construction d'expression génique spécifique d'un anti-virus. Cette population peut être combinée avec une population de lymphocytes CD4+ T ou une population de cellules progénitrices hématopoïétiques CD34+, et une ou plusieurs de ces populations cellulaires peuvent être distribuées à un individu positif à un virus autologue. L'invention concerne également des produits cellulaires thérapeutiques et des méthodes permettant de traiter des maladies, en particulier, des maladies infectieuses.
PCT/AU2003/001476 2002-11-07 2003-11-07 Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie WO2004042041A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2004548938A JP2006505259A (ja) 2002-11-07 2003-11-07 疾患特異的細胞傷害性tリンパ球の集団を産生および利用する手段
AU2003275767A AU2003275767A1 (en) 2002-11-07 2003-11-07 A means of producing and utilising a population of disease specific cytotoxic T-lymphocytes
US10/534,135 US20060051866A1 (en) 2002-11-07 2003-11-07 Means of producing and utilising a population of disease specific cytotoxic t-lymphocytes
CA002505379A CA2505379A1 (fr) 2002-11-07 2003-11-07 Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie
EP03810335A EP1558723A4 (fr) 2002-11-07 2003-11-07 Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US42451402P 2002-11-07 2002-11-07
US60/424,514 2002-11-07

Publications (1)

Publication Number Publication Date
WO2004042041A1 true WO2004042041A1 (fr) 2004-05-21

Family

ID=32312821

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2003/001476 WO2004042041A1 (fr) 2002-11-07 2003-11-07 Moyens de production et d'utilisation d'une population de lymphocytes t cytotoxiques specifiques d'une maladie

Country Status (6)

Country Link
US (1) US20060051866A1 (fr)
EP (1) EP1558723A4 (fr)
JP (1) JP2006505259A (fr)
AU (1) AU2003275767A1 (fr)
CA (1) CA2505379A1 (fr)
WO (1) WO2004042041A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010075061A3 (fr) * 2008-12-23 2010-08-12 Johnson & Johnson Research Pty Limited Traitement du sang
WO2020188573A1 (fr) * 2019-03-21 2020-09-24 Gamida-Cell Ltd. Fractions de cellules nk étendues appropriées pour une transplantation en polythérapie
US11723921B2 (en) 2011-05-26 2023-08-15 Geneius Biotechnology, Inc. Modulated immunodominance therapy

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1814580B1 (fr) 2004-11-24 2016-08-10 Fred Hutchinson Cancer Research Center Methodes d'utilisation d'il-21 pour l'immunotherapie adoptive et l'identification d'antigenes tumoraux
JP4719272B2 (ja) * 2005-11-22 2011-07-06 キオニクス,インコーポレイテッド 三軸加速度計

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999037313A1 (fr) * 1998-01-26 1999-07-29 Genzyme Corporation Hybrides de cellules effectrices immunes
WO2001094944A2 (fr) * 2000-06-02 2001-12-13 Memorial Sloan-Kettering Cancer Center Cellules presentatrices d'antigene artificiel et leurs methodes d'utilisation

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE60226853D1 (de) * 2001-02-20 2008-07-10 Ortho Mcneil Pharm Inc Zelltherapieverfahren für die behandlung von tumoren

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999037313A1 (fr) * 1998-01-26 1999-07-29 Genzyme Corporation Hybrides de cellules effectrices immunes
WO2001094944A2 (fr) * 2000-06-02 2001-12-13 Memorial Sloan-Kettering Cancer Center Cellules presentatrices d'antigene artificiel et leurs methodes d'utilisation

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
See also references of EP1558723A4 *
SUN L.Q. ET AL: "Resistance to human immunodeficiency virus I infection conferred by transaction of human peripheral blood lymphocytes with ribozyme, antisense, or polymeric trans-activation response elements", PROC NAT ACAD SCI USA, vol. 92, August 1995 (1995-08-01), pages 7272 - 7276, XP002041545 *
WEISSMAN D ET AL: "HIV Gag mRNA transfection of dendritic cells (DC) delivers encoded antigen to MHC class I and II molecules, causes DC maturation, and induces a potent human in vitro primary immune response", J IMMUNOL, vol. 165, no. 8, 2000, pages 4710 - 4717, XP008051732 *
ZARLING A ET AL: "Induction of primary human CD8+T lymphocyte responses in vitro using dendritic cells", J IMMUNOL, vol. 162, no. 9, 1999, pages 5197 - 5204, XP008051731 *
ZELING CAI ET AL: "Transfected Drosophila cells as a probe for defining the minimal requirements for stimulating unprimed CD8+ cells", PROC NAT ACAD SCI USA, vol. 93, no. 25, December 1996 (1996-12-01), pages 14736 - 14741, XP002205117 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010075061A3 (fr) * 2008-12-23 2010-08-12 Johnson & Johnson Research Pty Limited Traitement du sang
EP2392369A1 (fr) * 2008-12-23 2011-12-07 Therakos, Inc. Traitement du sang
US10926020B2 (en) 2008-12-23 2021-02-23 Mallinckrodt Hospital Products IP Limited Processing blood
US10940259B2 (en) 2008-12-23 2021-03-09 Mallinckrodt Hospital Products IP Limited Processing blood
US10953149B2 (en) 2008-12-23 2021-03-23 Mallinckrodt Hospital Products IP Limited Processing blood
US11723921B2 (en) 2011-05-26 2023-08-15 Geneius Biotechnology, Inc. Modulated immunodominance therapy
WO2020188573A1 (fr) * 2019-03-21 2020-09-24 Gamida-Cell Ltd. Fractions de cellules nk étendues appropriées pour une transplantation en polythérapie

Also Published As

Publication number Publication date
CA2505379A1 (fr) 2004-05-21
US20060051866A1 (en) 2006-03-09
EP1558723A4 (fr) 2006-02-15
EP1558723A1 (fr) 2005-08-03
JP2006505259A (ja) 2006-02-16
AU2003275767A1 (en) 2004-06-07

Similar Documents

Publication Publication Date Title
US20220265722A1 (en) Engineering and delivery of therapeutic compositions of freshley isolated cells
AU783429B2 (en) Veto cells effective in preventing graft rejection and devoid of graft versus host potential
US8722400B2 (en) Artificial antigen presenting cells and uses therefor
JPH08510134A (ja) Hiv感染およびエイズを対象としたリボザイム遺伝子治療
JP5805089B2 (ja) 細胞集団の製造方法
JP2022065022A (ja) 改変ヒト初代血液樹状細胞株を生成するための方法
MXPA04009287A (es) Proceso para producir linfocito citotoxico.
US20030082158A1 (en) Production of transduced hematopoietic progenitor cells
US20060051866A1 (en) Means of producing and utilising a population of disease specific cytotoxic t-lymphocytes
Ngok et al. Clinical gene therapy research utilizing ribozymes: application to the treatment of HIV/AIDS
Bollard et al. Gene marking studies of hematopoietic cells
EP4212618A1 (fr) Procédé de production de cellules t vdelta1+
AU2002320365A1 (en) Production of transduced hematopoietic progenitor cells
Varela-Rohena Redirection of T cell antigen specificity through genetic transfer of supraphysiologic T cell receptors
Spencer et al. Generating cytotoxic t lymphocytes against cervical carcinoma with lentivirus transduced antigen presenting cells
Ho et al. Ribozyme Gene Therapy Targeting Stem Cells for Human Immunodeficiency Virus Infection
MXPA94005978A (en) Methods for ex vivo therapy using antigen depressed cells charged with peptide for the application of

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2003275767

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2006051866

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2505379

Country of ref document: CA

Ref document number: 10534135

Country of ref document: US

Ref document number: 2004548938

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003810335

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003810335

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10534135

Country of ref document: US

WWW Wipo information: withdrawn in national office

Ref document number: 2003810335

Country of ref document: EP